1
|
Qi B, Wang Y, Zhu X, Gong Y, Jin J, Wu H, Man X, Liu F, Yao W, Gao J. miR-301a-mediated crosstalk between the Hedgehog and HIPPO/YAP signaling pathways promotes pancreatic cancer. Cancer Biol Ther 2025; 26:2457761. [PMID: 39846248 PMCID: PMC11760222 DOI: 10.1080/15384047.2025.2457761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its dismal prognosis and limited therapeutic options. In this study, we investigated the role of miR-301a in facilitating crosstalk between the Hedgehog (Hh) and HIPPO/YAP signaling pathways during the progression of PDAC. Our findings revealed that miR-301a served as a central regulatory node, targeting Gli1 within the Hh pathway and STK4 within the HIPPO/YAP pathway. Immunohistochemical and molecular analyses confirmed dysregulation of pathway components in pancreatic cancer, underscoring the pivotal role of miR-301a. Functional assays demonstrated the impact of miR-301a on cell proliferation and apoptosis, particularly in synergy with TNF-α. Overall, our study elucidated the intricate interplay between the Hh and HIPPO/YAP pathways mediated by miR-301a, providing valuable insights into potential therapeutic strategies for intervening in PDAC.
Collapse
Affiliation(s)
- Bing Qi
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
| | - Yuqiong Wang
- Department of Gastroenterology, The Hospital of 92608 People’s Liberation Army of China (PLA) Troops, Shanghai, China
| | - Xian Zhu
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Yanfang Gong
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jing Jin
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hongyu Wu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohua Man
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Feng Liu
- Department of Gastroenterology, Tongchuan People’s Hospital, Tongchuan, China
| | - Wenzhu Yao
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
| | - Jun Gao
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
2
|
Zhang P, Pan J, Lin S, Peng B, An C, Zhang J, Xu L, Lai Y, Yu H, Xu Z. Smart drug delivery platforms reprogramming cancer immune cycle to mitigate immune resistance of pancreatic tumors. Adv Drug Deliv Rev 2025; 224:115620. [DOI: 10.1016/j.addr.2025.115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
|
3
|
Malhotra P, Fyfe J, Emmanouilidi A, Casari I, Mellett NA, Huynh K, Pajic M, Greening DW, Meikle PJ, Falasca M. Oncogenic small extracellular vesicles enriched in sphingosine-1-phosphate play a crucial role in pancreatic cancer progression. Cell Signal 2025; 132:111775. [PMID: 40158707 DOI: 10.1016/j.cellsig.2025.111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Small extracellular vesicles (sEVs) from tumour cells mediate intercellular communication and signalling to regulate the progression of pancreatic ductal adenocarcinoma (PDAC). While we and others have shown that PDAC-derived sEVs comprise oncogenic protein and nucleic acid cargo, understanding the lipid landscape of these sEVs remains unknown. Lipids influence both the composition of sEVs and their roles in lipid metabolism and signalling pathways within the tumour microenvironment and tumorigenesis. We hypothesised that specific lipids in oncogenic sEVs might provide insights into PDAC. Comprehensive mass spectrometry-based lipidomic analysis was performed using liquid chromatography-electrospray ionisation-tandem mass spectrometry on sEVs isolated from PDAC and non-malignant pancreatic cell lines, patient-derived xenograft cell lines and plasma from the PDAC transgenic mouse model KPC (KRASWT/G12D/ TP53WT/R172H/Pdx1-Cre+/+). The sEV lipidomic analyses identified over 700 lipid species from 25 lipid classes and subclasses. Our results showed that, compared to non-malignant cells, PDAC-derived sEVs were enriched in specific lysophospholipids, particularly sphingosine-1-phosphate (S1P), a lipid known for its pivotal role in cancer pathogenesis. S1P enrichment was validated in plasma-derived sEVs from KPC mice compared to WT. To explore the functional implications of S1P enrichment, we conducted assays demonstrating that S1P in sEVs facilitated tubule formation in human microvascular endothelial cells and promoted cancer-associated fibroblast cell migration. We show that PDAC-derived sEVs are differentially enriched in specific lipids associated with cancer phenotype. Our findings highlight that PDAC-derived sEVs are enriched in specific lipids, particularly S1P, which plays a crucial role in promoting cancer progression.
Collapse
Affiliation(s)
- Pratibha Malhotra
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Jordan Fyfe
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Aikaterini Emmanouilidi
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Ilaria Casari
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Marina Pajic
- Translational Oncology Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David W Greening
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
4
|
Shi Y, Liu J, Cheng Q, Wu S, Song W, Wang K, Chen Z, Li X, Wei Q, Tayier D, Liao B, Yang Z. METTL3/IGF2BP3 mediates ORC6 via N6-methyladenosine modification to promote the progression of pancreatic ductal adenocarcinoma. Gene 2025; 955:149468. [PMID: 40185346 DOI: 10.1016/j.gene.2025.149468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is recognized globally as one of the most lethal tumours, and effective biomarkers to diagnose PDAC early are needed. ORC6, a subunit of the origin recognition complex (ORC), initiates DNA replication and ensures genomic stability. Previous studies have indicated that ORC6 is procarcinogenic in various cancers, yet its role in PDAC remains uninvestigated. METHODS We evaluated the relationships between ORC6 expression and the clinical features of patients with PDAC with the TCGA, GTEx, and GEO databases. The role of ORC6 in PDAC cells was explored by RNA interference in vitro and in vivo. Next, we verified the effect of the METTL3/IGF2BP3/ORC6 axis on PDAC progression by western blotting, RT-qPCR, RNA immunoprecipitation, and methylated RNA immunoprecipitation. Finally, transcriptome analysis was performed to explore the influence of ORC6 on p53 in PDAC cells. RESULTS Elevated ORC6 levels were observed in PDAC cells, which correlated with poorer clinical outcomes. Both in vivo and in vitro experiments demonstrated that ORC6 knockdown suppressed proliferation and promoted apoptosis. Additionally, we demonstrated that METTL3/IGF2BP3 interacted with ORC6 mRNA via N6-methyladenosine modification to improve ORC6 mRNA stability. Transcriptomic analysis and experiments indicated that ORC6 promoted PDAC progression by inhibiting serine-15 phosphorylation in p53. CONCLUSION Our findings validate the role of ORC6 in PDAC and support the hypothesis that the METTL3/IGF2BP3/ORC6/p53 axis may be a novel therapeutic target for PDAC, and inhibiting this axis may be an advantageous therapeutic strategy for curing PDAC.
Collapse
Affiliation(s)
- Yang Shi
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Junwei Liu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China
| | - Qian Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Shuaihui Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Wenjing Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Kunlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Zhinan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Xinyin Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Qifeng Wei
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Dilinigeer Tayier
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Bo Liao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China.
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China.
| |
Collapse
|
5
|
Sun G, Wu Y, Li J, Yang M, Xu H, Li Y, Tong P, Shao R, Liu Y, Kong X. Quercetin liposomes conjugated with hyaluronidase: An efficient drug delivery system to block pancreatic cancer. J Control Release 2025; 382:113642. [PMID: 40127723 DOI: 10.1016/j.jconrel.2025.113642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic cancer characterized with intense hydraulic tissue in tumor extracellular matrix (ECM) resists most of chemotherapeutic drugs. Increased levels of hyaluronic acid (HA) represent the primary component of the hydraulic tissue, rendering tumors protective from drug targeting. Quercetin (Que), a natural flavonoid, has the ability to inhibit tumor cell growth in a number of cancers; however, its poor water solubility and low bioavailability largely limit its application in cancer therapy. Hence, we developed an efficient drug delivery system by encapsulation of Que. into liposomes and conjugation with hyaluronidase (HAase) at liposome surface, termed as HQL. In the presence of HAase, HQL were predominantly accumulated at tumor with enhanced permeability and retention effect. Treatment of xenografted tumor mice with HQL gave rise to suppressed tumor growth, while no toxic effects were observed in mice. HQL demonstrated the strong ability to inhibit cell proliferation, promote cell apoptosis, and induce arrest at G2/M cell cycle in pancreatic cancer lines, three-dimensional cultured cell spheroids and pancreatic ductal adenocarcinoma (PDAC)-derived organoids. Mechanistically, HQL downregulated expression of cell cycle-associated protein (CCNB1, CDK1 and PLK1) and cell apoptosis-associated factors PI3K/AKT and Bcl-2. In summary, HQL degraded HA in the tumor microenvironment to enhance nano-particle penetration and inhibited tumor cell growth, eliciting efficacy of anti-tumor therapy. Thereof, HQL may provide a novel efficient drug delivery approach for the adjuvant treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ge Sun
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Ying Wu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Jiekai Li
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hang Xu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Yiping Li
- Department of Oncology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430065, China
| | - Peilin Tong
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China; Shanghai Key Laboratory of Biliary Tract Diseases, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of General Surgery, Jiading Branch, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China.
| | - Xianming Kong
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| |
Collapse
|
6
|
Imaoka H, Ikeda M, Ozaka M, Oshima K, Okano N, Shimizu S, Tsumura H, Komatsu Y, Yamashita T, Kataoka S, Nagano H, Hisano T, Sasaki M, Kobayashi S, Fukushima T, Mitsunaga S, Furukawa T, Hamauchi S, Ueno M, Furuse J. Phase 1/2 study of liposomal irinotecan plus S-1 for metastatic pancreatic cancer refractory to gemcitabine-based treatment. Eur J Cancer 2025; 222:115424. [PMID: 40252631 DOI: 10.1016/j.ejca.2025.115424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) improves survival in gemcitabine-refractory metastatic pancreatic cancer (PC) but requires a central venous port. S-1, an oral fluoropyrimidine with proven efficacy in PC, may replace 5-FU/LV in nal-IRI plus 5-FU/LV, potentially enhancing both convenience and antitumor effect. METHODS This single-arm, open-label, phase 1/2 study included patients with histologically or cytologically confirmed adenocarcinoma, aged 20-80 years, an Eastern Cooperative Oncology Group performance status of 0-1, with metastatic disease, and refractory to gemcitabine-based treatment. The primary endpoint in phase 1 part was the frequency of dose-limiting toxicity (DLT) to nal-IRI plus S-1. The primary endpoint in phase 2 part was overall survival. This trial was registered in the Japan Registry of Clinical Trials database (jRCTs031210040). RESULTS In phase 1 part, one patient with DLT was observed at nal-IRI 70 mg/m2 (day 1) with S-1 80 mg/m2/day (day 1-7) in a 2-week cycle, establishing this as the recommended phase 2 dose (RP2D). Forty-nine patients from phase 1 (n = 6) and phase 2 part (n = 43) were treated with the RP2D, and their results were pooled. Median overall survival was 10.3 months (95 % confidence interval, 8.1-12.0 months). A confirmed partial response was achieved in 10 patients (20.4 %). The most frequent treatment-emergent adverse events were hypoalbuminemia (98.0 %), anemia (98.0 %), and anorexia (81.6 %). There were no treatment-related deaths. CONCLUSIONS This study demonstrated that nal-IRI plus S-1 exhibited promising efficacy and an acceptable safety profile in patients with metastatic PC refractory to gemcitabine-based treatment.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kotoe Oshima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Terumasa Hisano
- Department of Hepato-Biliary-Pancreatology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Hamauchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
7
|
Zhang Z, Liu Z, Yao Y, Li M, Shen C, Zhou F. Exploring the clinical significance of TPX2 in pancreatic cancer: from biomarker to immunotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6843-6864. [PMID: 39688710 DOI: 10.1007/s00210-024-03628-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024]
Abstract
Pancreatic cancer (PC) is a highly aggressive malignancy characterized by a dismal prognosis. The present study is designed to elucidate the pivotal role of Xenopus kinesin-like protein 2 (TPX2) as a biomarker with substantial clinical prognostic significance in PC. By conducting a comprehensive analysis of RNA sequencing data and protein expression profiles obtained from multiple databases, we observed a pronounced upregulation of TPX2 expression in PC tissues compared to normal pancreatic tissues. Importantly, TPX2 emerged as an independent prognostic factor, demonstrating remarkable diagnostic accuracy. Notably, its expression levels were found to be significantly associated with the PC immune microenvironment and sensitivity to various therapeutic modalities. Functional assays revealed that the silencing of TPX2 markedly inhibited PC cell proliferation, metastasis, and the growth of subcutaneous tumors in PC mouse models. These effects were potentially mediated by the activation of CD8+ T cell immune responses and the inhibition of cell cycle progression and adhesion mechanisms. Taken together, our findings indicate that TPX2 may serve as a critical biomarker for the diagnosis and clinical management of patients with PC.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Zixian Liu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Ying Yao
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Cunsi Shen
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| |
Collapse
|
8
|
Gao X, Zhang G, Wang F, Ruan W, Sun S, Zhang Q, Liu X. Emerging roles of EGFL family members in neoplastic diseases: Molecular mechanisms and targeted therapies. Biochem Pharmacol 2025; 236:116847. [PMID: 40044051 DOI: 10.1016/j.bcp.2025.116847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Epidermal growth factor-like proteins (EGFLs) contain more than a single EGF/EGF-like domain within their protein structure. To date, ten EGFL family members (EGFL1-10) have been characterized across diverse tissues and developmental stages under different conditions. In this review, we conclude that EGFLs are instrumental in regulating biological activities and pathological processes. Under physiological conditions, EGFLs participate in angiogenesis, neurogenesis, osteogenesis, and other processes. Under pathological conditions, EGFLs are linked with different diseases, particularly cancers. Furthermore, we highlight recent advancements in the study of EGFLs in biological conditions and cancers. In addition, the regulatory role and key underlying mechanism of EGFLs in mediating tumorigenesis are discussed. This paper also examines potential antagonists that target EGFL family members in cancer therapeutics. In summary, this comprehensive review elucidates the critical role of EGFLs in neoplastic diseases and highlights their potential as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Guopeng Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Wenhui Ruan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China.
| |
Collapse
|
9
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
10
|
Li Z, Duan J, Liu Z, Li W, Mai Y, Fu H, Yuan G, Wang J. A triple-mode strategy on JQ1-loaded nanoplatform for superior antitumor therapy in pancreatic cancer. Mater Today Bio 2025; 32:101696. [PMID: 40225138 PMCID: PMC11986615 DOI: 10.1016/j.mtbio.2025.101696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic cancer's dire prognosis urgently calls for innovative therapeutic strategies. JQ1, a bromodomain 4 inhibitor, exhibits potent anti-tumor activity in preclinical models but faces limitations due to rapid resistance development. Here, we developed a novel multifunctional nanoplatform, JQ1@MSN/FeTA-iRGD, which implemented a triple-mode strategy integrating apoptosis, ferroptosis, and immunogenic cell death for optimized treatment of pancreatic cancer. The particles could precisely target tumors in mice and achieve efficient release of JQ1 and Fe2+ through internalization in the acidic tumor environment. The nanoplatform amplified reactive oxygen species and mitochondrial damage to disrupt the redox homeostasis, thus synergistically escalating apoptosis and ferroptosis for the destruction of tumor cells, circumventing the rapid drug resistance associated with monotherapy. Meanwhile, dying cancer cells released damage-associated molecular patterns, which facilitated immunogenic cell death and triggered antitumor immune responses, guaranteeing the sustained efficacy of the treatment. Moreover, the system exhibited favorable biocompatibility, supporting its feasibility for clinical translation. Our results demonstrated that this novel strategy, combining apoptosis, ferroptosis, and immunogenic cell death, overcame the limitations of monotherapy with JQ1, providing a superior, targeted, and sustainable treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Zhiguo Li
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jinxin Duan
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhiwen Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weifan Li
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yiyin Mai
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hao Fu
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Guotao Yuan
- College of Chemistry and Environmental Engineering, Shenzhen University, 518060, China
- Department of Otolaryngology, Longgang E.N.T. Hospital & Shenzhen Key Laboratory of E.N.T., Shenzhen, 518116, China
| | - Jiawei Wang
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| |
Collapse
|
11
|
Zhou X, Ling Y, Huang L, Yang F, Zhang Y, Lan Y. HIF-3α Facilitates the Proliferation and Migration in Pancreatic Cancer by Inhibiting Autophagy Through Downregulating TP53INP2. Cell Biochem Biophys 2025; 83:2139-2150. [PMID: 39614944 DOI: 10.1007/s12013-024-01624-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 05/20/2025]
Abstract
Pancreatic cancer is a highly aggressive malignant tumor, often diagnosed late, leading to a poor prognosis and extremely high mortality rates. In recent years, the role of cellular autophagy in tumors has become increasingly prominent, gradually becoming an important target for malignant tumors. HIF-3α is a member of HIF family with potential oncogenic function. However, the role of HIF-3α in pancreatic cancer is not clear. The present study revealed its role in pancreatic cancer by exploring the regulatory mechanism of HIF-3α on autophagy. HIF-3α was found markedly upregulated in pancreatic cancer cell lines. In HIF-3α silenced MiaPaCa-2 cells, largely declined migration distance, reduced number of invaded cells and colonies, increased number of autophagosome, downregulated p62, and upregulated Beclin1, LC3II/I, and ATG7 were observed, accompanied by elevated TP53INP2 expressions. on the contrary, in HIF-3α overexpressed PANC-1 cells, notably increased migration distance, and elevated number of invaded cells and colonies were observed, along with decreased autophagosome, upregulated p62, and downregulated Beclin1, LC3II/I, ATG7, and TP53INP2. Subsequently, HIF-3α overexpressed PANC-1 cells were transfected with TP53INP2 overexpressing vector. The influence of HIF-3α overexpression on the proliferation, migration, invasion, and autophagy was abolished by TP53INP2 overexpressing. Furthermore, HIF-3α overexpression facilitated the in vivo growth of PANC-1 cells, accompanied by the autophagy inhibition in tumor tissues, which were remarkably abolished by TP53INP2 overexpressing. Collectively, HIF-3α facilitated the proliferation and migration in pancreatic cancer by inhibiting autophagy through downregulating TP53INP2.
Collapse
Affiliation(s)
- Xianfei Zhou
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China
| | - Yisheng Ling
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China
| | - Luoshun Huang
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China
| | - Fan Yang
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China
| | - Yang Zhang
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China
| | - Yong Lan
- Department of hepatobiliary surgery, Taizhou Municipal Hospital, No. 581, Shifu Avenue East, Jiaojiang District, Taizhou City, 318000, Zhejiang, China.
| |
Collapse
|
12
|
Zhan Y, Zhang K, Fan Y, Lin S, Wu J, Xu H. Lipids, lipid-lowering drug target genes and pancreatic cancer: a Mendelian randomization study. Int J Clin Pharm 2025; 47:747-754. [PMID: 39821006 DOI: 10.1007/s11096-025-01866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a malignant tumor with a low survival rate. Lipid modifiers show potential for PC therapy, but evidence is lacking. AIM This Mendelian randomization (MR) study aimed to explore the relationship between lipid traits, and lipid-lowering drug target genes with PC risk. METHOD Genetic instrumental variables associated with lipid traits and lipid-lowering drug target genes were used to perform MR analyses of PC risk. MR estimation was based on genome-wide association study data from two large sample sets, and the MR results were meta-analyzed to assess their impact on PC risk. To ensure the reliability of lipid-modifying drug targets, we conducted a Summary Data-based Mendelian Randomization (SMR) analysis. Additionally, a two-step MR analysis was employed to explore potential mediating effects. RESULTS In two independent datasets, HMG-CoA reductase (HMGCR) inhibition was statistically associated with a lower risk of PC (OR 0.50, [95% CI 0.25-1.00]; p = 0.0453). The results were further supported by SMR analysis, which showed a similar association (OR 0.51, [95% CI 0.28-0.96]; p = 0.0369). Mediation analysis revealed that 11.69% of the protective effect of HMGCR inhibitors on PC is mediated through lower BMI levels. No significant effect of lipid traits and the other eight lipid-lowering drug targets on PC risk was found. CONCLUSION This study suggests that HMGCR may be a potential drug target for the treatment or prevention of PC, providing important insights into the use of lipid-targeted drugs in PC therapy.
Collapse
Affiliation(s)
- Yuxuan Zhan
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Kai Zhang
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Yiqun Fan
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyi Lin
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Jian Wu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Hongxia Xu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Zaki-Dizaji M, Taheri Z, Heiat M, Hushmandi K. Tumor-educated platelet, a potential liquid biopsy biosource in pancreatic cancer: A review. Pathol Res Pract 2025; 270:155986. [PMID: 40286788 DOI: 10.1016/j.prp.2025.155986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 04/13/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic cancer (PC) is a frequent and aggressive digestive system cancer with a very poor prognosis. The best chance for recovery lies in early surgical removal of the tumor. Unfortunately, because PC often develops without noticeable symptoms, diagnosis is frequently delayed. Limited treatment options, the metastasis potential of pancreatic cancer cells, and its generally poor prognosis mean that patients are often diagnosed late, significantly reducing the effectiveness of treatment. Consequently, there's a critical need for new biomarkers and technologies to improve early detection through screening. Recently, the liquid biopsy has developed as a powerful means for detecting and monitoring cancer at the molecular level. Its advantages include the ease and non-invasive nature of sample collection and its ability to reflect the dynamic changes within a tumor. Platelets, the second most numerous type of blood cell, offer a particularly promising source for liquid biopsy. It is known that cancer affects various aspects of platelets, including their number, size, activation state, and the proteins and RNA they contain. However, the full implications of these changes for cancer detection have not yet been fully integrated into routine clinical practice. Platelets have a unique ability to captivate nucleic acids and proteins from their surroundings, and they alter their transcriptome in response to external signals. This leads to the development of tumor-educated platelets (TEPs). Liquid biopsies that utilize TEP biomarkers hold considerable potential for screening, early detection, prognosis, guiding personalized treatment strategies, ongoing monitoring of the disease, and predicting recurrence. Encouraging results from preclinical studies have highlighted the potential of platelets as a novel liquid biopsy source for a wide range of cancers. This review will explore the potential of using platelets as a liquid biopsy method, specifically for pancreatic cancer.
Collapse
Affiliation(s)
- Majid Zaki-Dizaji
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Taheri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Zhang J, Tan Q, Fan Y, Xiao L, Zheng Z, Li K, Jing W, Song H, Liu X, Tan C, Wang X. Non-hypervascular pancreatic neuroendocrine neoplasms differentiation from CA19-9 negative pancreatic ductal adenocarcinomas based on contrast CT: A large sample series. Eur J Radiol 2025; 187:112095. [PMID: 40209484 DOI: 10.1016/j.ejrad.2025.112095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
PROPOSE This study aims to evaluate the effectiveness of contrast-enhanced computed tomography (CT) in distinguishing non-hypervascular pancreatic neuroendocrine neoplasms (PNENs) from pancreatic ductal adenocarcinomas (PDACs) with a normal serum level of carbohydrate antigen 19-9 (CA19-9) levels. METHODS This retrospective study included 134 patients with pathologically confirmed non-hypervascular PNENs and 128 patients with CA19-9-negative PDACs, all of whom underwent contrast-enhanced CT prior to surgery between January 2015 and March 2024. Following independent evaluation by two radiologists, qualitative features from both groups were extracted in the arterial and portal venous phase and subsequently compared using univariate and multivariate analysis. RESULTS Patients with CA19-9 negative PDACs were significantly older than those with non-hypervascular PNENs (p < 0.001), and the majority of PDACs were located in the head of the pancreas (p < 0.01).Univariate analysis showed that non-hypervascular PNENs exhibited a higher frequency of well-defined tumor margins (p < 0.001) and calcification (p = 0.032) and a lower frequency of local invasion (p < 0.001), peripancreatic vascular invasion (p = 0.001), intra- or extrahepatic bile duct dilatation (p < 0.001), distal main pancreatic duct dilatation (p < 0.001), regional lymphadenopathy (p < 0.001) and tumor homogeneity (p < 0.001) when compared to CA19-9 negative PDACs. Multivariate analysis identified the absence of local invasion (Odds Ratio (OR) = 0.233; 95 % Confidence Internals (95 % CI):0.114-0.476; p < 0.001), absence of peripancreatic vascular invasion (OR = 0.434; 95 % CI:0.217-0.870; p = 0.019), a normal distal main pancreatic duct diameter (OR = 0.398; 95 % CI:0.202-0.785; p = 0.008), absence of regional lymphadenopathy (OR = 0.455; 95 % CI:0.238-0.870; p = 0.017) and tumor heterogeneity (OR = 0.240; 95 % CI:0.126-0.456; p < 0.001) as significant predictors of non-hypervascular PNENs. The area under the receiver operating characteristic curve for the radiological feature model was 0.829 based on logistic regression. CONCLUSIONS Qualitative features in contrast-enhanced CT images could be beneficial in differentially diagnosing non-hypervascular PNENs and CA19-9 negative PDACs.
Collapse
Affiliation(s)
- Jinyin Zhang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingquan Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Fan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Liu Xiao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenyi Jing
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haiyu Song
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan Province, China
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Xing Wang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
15
|
Shi H, Liu X, Xing C, Guo S, Zheng Y, Tan W, Ge Y, Xu J, Li Y, Song J. DNMT1-Induced Downregulation of CBX7 Inhibits ERK Phosphorylation and Promotes Pancreatic Ductal Adenocarcinoma Progression. FASEB J 2025; 39:e70571. [PMID: 40387566 PMCID: PMC12087528 DOI: 10.1096/fj.202402903r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/25/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancer types, characterized by an alarmingly low 5-year survival rate. DNA methylation has been implicated in the progression of various tumors, with DNA methyltransferase 1 (DNMT1) being the most extensively studied enzyme in this context. However, the expression patterns and underlying mechanisms of DNMT1 in PDAC remain poorly understood. The levels of DNMT1 and CBX7 in PDAC tissues and cells were determined by IHC and Western blot. ChIP and dual-luciferase reporter assays confirmed the interaction between DNMT1 and the CBX7 promoter. Cellular functions were evaluated through CCK-8, wound healing, and transwell assays. The expression of MAPK-related proteins was analyzed by Western blot. DNMT1 expression was upregulated in PDAC tissues and cell lines, whereas CBX7 expression was downregulated. Silencing DNMT1 inhibited cell proliferation, migration, and invasion in PDAC by modulating CBX7 expression. Moreover, DNMT1 methylates the CBX7 promoter region, leading to increased ERK phosphorylation, which subsequently drives tumorigenesis and metastasis in PDAC. DNMT1 promotes the malignant progression of PDAC through the CBX7/ERK pathway. Our study provides evidence for potential therapeutic targets for the comprehensive treatment of PDAC.
Collapse
Affiliation(s)
- Haowei Shi
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Xu Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Cheng Xing
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Shiqi Guo
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yangyang Zheng
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Wendan Tan
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yunpeng Ge
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Jingyong Xu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yao Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Jinghai Song
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
16
|
Shi W, Tang H, Tan S, Wang L, Yu Z, Gao S, Zhou J. MMA-induced LOXL2 + PSCs promote linear ECM alignment in the aging pancreas leading to pancreatic cancer progression. Cell Death Dis 2025; 16:419. [PMID: 40425551 PMCID: PMC12116754 DOI: 10.1038/s41419-025-07751-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 05/02/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an age-associated malignancy closely linked to the extracellular matrix (ECM). However, the impact of age-related ECM changes in the normal pancreas on PDAC progression remains unclear. Here, we find that increased linear ECM alignment in normal pancreatic tissues from aged PDAC patients is associated with PDAC progression and worse outcomes. Furthermore, serum methylmalonic acid (MMA) levels are elevated in aged PDAC patients and associated with increased linear ECM alignment in normal pancreatic tissues of PDAC patients. Functionally, MMA promotes LOXL2 expression in pancreatic stellate cells (PSCs), increases linear ECM alignment in normal pancreatic tissues, and facilitates tumor progression. Mechanistically, MMA upregulates KLF10, which forms a transcriptional complex with SP1 to enhance LOXL2 expression in PSCs. Our study demonstrates the role of MMA-induced LOXL2+PSCs in ECM remodeling, thus serving as a potential therapeutic target to mitigate PDAC progression in aged patients. Schematic diagram showing the molecular mechanism by which MMA-induced LOXL+PSCs promote PDAC progression in the aging pancreas. In aged individuals, elevated levels of MMA in the blood induce the activation of the KLF10/SP1‒LOXL2 axis in PSCs to increase linear ECM alignment. Following the initiation of pancreatic cancer, this increased linear ECM alignment leads to increased tumor invasion into surrounding tissues, resulting in a greater proportion of stage T3/T4 tumors and a greater incidence of LVI and PNI in aged patients, ultimately leading to poorer outcomes (This schematic was created with www.figdraw.com ,export id: PRPRS4e268).
Collapse
Affiliation(s)
- Wenyuan Shi
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Haodong Tang
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Siyuan Tan
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Lishan Wang
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Zeqian Yu
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiahua Zhou
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China.
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
17
|
Xu LL, Wang M, Wang YK, Chen YJ, Zhang YX, Zhang YQ, Cheng SB, Xie M, Huang WH. Vessel-Like Microtunnels with Biomimetic Octopus Tentacles for Seizing and Detecting Exosomes to Diagnose Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2502763. [PMID: 40424013 DOI: 10.1002/smll.202502763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/17/2025] [Indexed: 05/28/2025]
Abstract
Microchip-based exosome analysis has emerged as a promising approach for liquid biopsy in cancer diagnosis, treatment monitoring, and prognostic evaluation. However, current microchips for exosome analysis typically rely on planar, 2D channel structures with affinity properties, which require complex fabrication but deliver suboptimal separation and detection performance. This study presents a novel vessel-like microtunnel chip, integrated with biomimetic octopus tentacles, achieving an exosome isolation efficiency of 90.4%. The innovative design incorporates interwoven, 3D micropathways, enhancing fluid dynamics and promoting efficient mixing between exosomes and microchannels. Nanofiber-coated silicon microspheres, functionalized with synthetic peptides, mimic octopus tentacles to anchor the microtunnels, dynamically extending under fluid shear forces to specifically recognize lipid bilayer structures for exosome capture. This platform incorporates enzyme-catalyzed signal amplification using Au nanoprobes for colorimetric detection to sensitively analyze four protein markers on plasma-derived exosomes from 60 clinical samples. Machine learning is used to develop a diagnostic model, achieving an area under the curve (AUC) of 0.9888 in distinguishing pancreatic cancer from pancreatitis and healthy controls. This approach provides a rapid, sensitive, accurate, and user-friendly method for pancreatic cancer diagnosis, addressing the clinical challenges of early detection and the frequent misdiagnosis of pancreatic cancer as pancreatitis.
Collapse
Affiliation(s)
- Li-Li Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Ming Wang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Yi-Ke Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Yi-Jing Chen
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Yu-Xin Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Yan-Qiu Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Shi-Bo Cheng
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjia Lake West Road, Wuhan, 430065, P. R. China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| |
Collapse
|
18
|
Li G, Zhang Y, Wang J, Liang Y, Zhang Y, Chi Y, Sun C, Hu Z, Wu S, Lu GL, Wen J, Zhang Z. Phosphatidylcholine Chain-Length of Bioinspired Lipoprotein Modulates Interactions with Collagen for Intratumor Delivery in Pancreatic Cancer. ACS NANO 2025; 19:19126-19140. [PMID: 40375717 DOI: 10.1021/acsnano.4c18963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Pancreatic cancer, one of the most lethal malignant tumors, is greatly challenged by poor drug delivery efficiency because of the dense and intricate desmoplastic stroma. Given the abnormal expression of scavenger receptor B type 1 (SR-B1) and the dense collagen I (COL1)-enriched extracellular matrix (ECM) barrier in pancreatic tumors, we developed four BLPs with regular chain-length of phosphatidylcholine (PC) (BLP-M, BLP-S, BLP-P, and BLP-H) to enhance their intratumoral delivery for chemotherapy. With the increase of PC chain-length in BLPs, these BLPs exhibited regular tendency of increased affinity to COL1, reduced diffusion capacity in COL1 hydrogel, lessened tumor accumulation and intratumor distribution, and declined efficacy of prolonging survival in the orthotopic pancreatic cancer model. Particularly, the DMPC-based BLP-M system showed the lowest affinity to COL1 and the highest diffusion capacity in the COL1-based ECM barrier, thereby causing the best efficacy of specific tumor accumulation, intratumor delivery, and survival prolongation in an orthotopic pancreatic tumor model. Thereby, this study provided substantial insights into the targeting and intratumor delivery in pancreatic cancer, and DMPC-based BLPs represented an encouraging delivery platform for effective cancer chemotherapy.
Collapse
Affiliation(s)
- Guodong Li
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yichen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Jinhua Wang
- The First People's Hospital of Taian, Shandong 271000, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yuan Zhang
- The First People's Hospital of Taian, Shandong 271000, China
| | - Yifei Chi
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Carter Sun
- The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zixin Hu
- Artificial Intelligence Innovation and Incubation Institute of Fudan University & Shanghai Academy of Artificial Intelligence for Science, Shanghai 201111, China
| | - Shiyang Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Guo-Liang Lu
- The University of Auckland, Auckland 1142, New Zealand
| | - Jingyuan Wen
- The University of Auckland, Auckland 1142, New Zealand
| | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| |
Collapse
|
19
|
Song N, Tao R, Li H, Zhang R, Huang Y, Zhang L, Liu Y, Yang D, Yao C. Spatially Controlled Co-Delivery of Diagnostic and Therapeutic Agents Using DNA Nanoframeworks for Pancreatic Cancer Precision Therapy. Angew Chem Int Ed Engl 2025; 64:e202500566. [PMID: 40123438 DOI: 10.1002/anie.202500566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Theranostic platforms that integrate diagnostic and therapeutic functionalities offer promising strategies for precision medicine, particularly in the treatment of major diseases. However, the development of platforms capable of achieving spatially controlled detection and therapy at the lesion site remains a significant challenge. Herein, we present a dual-stimuli-responsive DNA nanoframework that achieve spatially controlled codelivery of molecular beacon (MB) and Cas9 ribonucleoprotein (RNP), enabling simultaneous specific optical detection and efficient gene therapy for pancreatic cancer. The DNA nanoframeworks are synthesized via precipitation polymerization, utilizing acrylamide-modified DNA to initiate a hybridization chain reaction that facilitates the effective loading of MB-extended and sgRNA-conjugated DNA hairpins. The Cas9 protein is efficiently loaded into the nanoframeworks through phase transition-induced polymer chain rearrangement, overcoming steric hindrance. Upon aptamer-mediated internalization into PANC-1 cells, the overexpressed apurinic/apyrimidinic endonuclease 1 and ribonuclease H in cancer cells induce site-specific cleave of MB and DNA-RNA hybrid duplex, respectively. This cleavage restores fluorescence for specific optical detection, whereas the released Cas9 RNP performs gene editing for efficient therapy. Low fluorescence background and favorable biocompatibility are observed in normal cells. In a pancreatic cancer mouse model, the platform demonstrates significant detection-guided antitumor efficacy, highlighting its potential for precision medicine in cancer therapy.
Collapse
Affiliation(s)
- Nachuan Song
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Ruoyu Tao
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
| | - Hongjin Li
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
| | - Rui Zhang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Yan Huang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
| | - Le Zhang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
| | - Dayong Yang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Chi Yao
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin, 300350, P.R. China
| |
Collapse
|
20
|
Liu J, Gong Y, Zeng X, He M, He B, Gao W, Gao Y. Enhanced anti-cancer effect of AMTB hydrochloride via chitosan nanoparticles in pancreatic cancer. BMC Cancer 2025; 25:944. [PMID: 40420025 PMCID: PMC12105253 DOI: 10.1186/s12885-025-14356-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Pancreatic cancer is a malignancy with poor prognosis and high mortality. This study investigated the use of chitosan nanoparticles (CS-NPs) to encapsulate AMTB, a TRPM8 inhibitor, as a novel strategy to enhance therapeutic efficacy in pancreatic cancer. TRPM8 was overexpressed in pancreatic cancer tissues and associated with poor patient prognosis. AMTB inhibited pancreatic cancer cell proliferation, migration, and invasion by suppressing the EMT process and MMP2/9 expression. CS-NPs@AMTB were successfully synthesized, exhibiting excellent drug release profiles and stronger anti-tumor effects than free AMTB. Both AMTB and CS-NPs@AMTB demonstrated favorable biological safety. This is the first study to apply chitosan nanoparticles for AMTB delivery in pancreatic cancer, significantly enhancing its anti-tumor and anti-metastatic effects (ahout 70% reduction in tumor size). These findings suggest that CS-NPs@AMTB might overcome current therapeutic limitations by improving drug efficacy and targeting metastasis in pancreatic cancer. With further validation through preclinical and clinical studies, this nanoparticle-based delivery strategy holds promise for integration into future therapeutic regimens and personalized treatment approaches.
Collapse
Affiliation(s)
- Jiefeng Liu
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China.
| | - Yujing Gong
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| | - Xinyu Zeng
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| | - Miao He
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| | - Bin He
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| | - Wenbin Gao
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| | - Yong Gao
- Department of General Surgery, Changsha Hospital Affiliated to Hunan Normal University, The Fourth Hospital of Changsha, No. 200 Jinxing Road, Wangcheng District, Changsha, 410219, Hunan, China
| |
Collapse
|
21
|
Qi J, Fang C, Deng C, Shi F, Yao Q. Mesoporous Magnetic Graphene for Serum Metabolic Profiling in Non-Invasive Early Detection and Diagnosis of Pancreatic Ductal Adenocarcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29995-30005. [PMID: 40355809 DOI: 10.1021/acsami.5c03176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal cancer, typically diagnosed at advanced stages due to its asymptomatic onset and challenges in early detection. To address the critical need for the early diagnosis of PDAC, we developed a laser desorption/ionization mass spectrometry (LDI-MS) platform based on mesoporous silica-modified magnetic graphene (MG@mSiO2). MG@mSiO2 exhibited exceptional ultraviolet (UV) absorption, efficient ionization, and minimal background interference, enabling high-resolution profiling of serum metabolic fingerprints (SMFs). Based on the extracted SMFs, we constructed a Random Forest (RF) model to classify PDAC patients, high-risk (HR) individuals, and healthy controls (HC), achieving an accuracy of 97.5% in the independent test set. Additionally, a six-metabolite biomarker panel was identified, showing strong diagnostic potential with sensitivity and accuracy exceeding 89.1% for distinguishing HC from PDAC. When coupled with the serological marker carbohydrate antigen 19-9 (CA19-9), the integrated strategy delivered significantly improved diagnostic performance, achieving high accuracy ranging from 95.3% to 100% in distinguishing HR and PDAC patients from HC. Furthermore, metabolic pathway analysis revealed key pathways associated with PDAC progression, providing mechanistic insights into the disease. This work provides a powerful diagnostic tool for PDAC screening, establishing a foundation for early detection and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Jia Qi
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Caiyun Fang
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Chunhui Deng
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Fangying Shi
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| |
Collapse
|
22
|
Xu D, Lv N, Wang Q, Wu Y, Zhang K, Miao Y, Wei J, Tu M, Jiang K. The feasibility and potential benefits of administering adjuvant chemotherapy in resected pancreatic cancer patients unable to promptly remove intraperitoneal drainage post-surgery: a retrospective cohort study. BMC Cancer 2025; 25:901. [PMID: 40394527 DOI: 10.1186/s12885-025-14262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025] Open
Abstract
OBJECTIVE Pancreatectomy remains associated with a high incidence of complications. In certain cases, patients with pancreatic ductal adenocarcinoma (PDAC) face challenges in removing intraperitoneal drainage after surgery, leading to potential delays in the administration of adjuvant chemotherapy (AC) and potentially impacting survival outcomes. The objective of this study was to evaluate the feasibility and potential benefits of AC in PDAC patients who are unable to remove intraperitoneal drainage over 30 days. METHODS Between January 2021 and December 2022, a total of 220 patients with resected PDAC received AC at our center. Among them, 84 patients experienced persistent intraperitoneal drainage lasting more than 30 days postoperatively. Of these, 38 patients (45.2%) initiated AC despite the ongoing presence of drainage and were classified as the AC(d+) group, while the remaining 46 patients (54.8%) began AC only after successful drainage removal, and were categorized as the AC(d-) group. The other 136 patients, who underwent prompt removal of intraperitoneal drainage, were assigned to the AC(pr) group. Baseline information, surgery-related outcomes, and chemotherapy-related adverse events were collected and compared between the two groups, and factors that affected recurrence-free survival (RFS) were also analysed. RESULTS Of the 220 patients included in the study, 107 (48.7%) experienced grade 3-4 chemotherapy-related adverse events. The interval from surgery to the initiation of AC was similar between the AC(d+) and AC(pr) groups (50 vs. 57 days, P = 0.108). However, it was significantly shorter in the AC(d+) group compared to the AC(d-) group (50 vs. 61 days, P = 0.015). Notably, no additional chemotherapy-related adverse events were observed in the AC(d+) group compared to either the AC(d-) or AC(pr) groups. The estimated 1-year and 2-year survival rates were 85.6% and 60.5%, respectively, for the AC(d-) group, and 95.8% and 61.0% for the AC(d+) group. In the AC(pr) group, the corresponding survival rates were 89.1% and 64.0%. Cox multivariate regression analysis demonstrated that tumour grade differentiation, completed six cycles of therapy, the interval from surgery to the initiation of AC and resection margins were independent factors affecting RFS. CONCLUSION Administering AC was safe for patients who underwent resection for PDAC and encountered challenges in the prompt removal of intraperitoneal drainage beyond 30 days post-surgery. The proactive management of preventing delays in chemotherapy administration could reduce the early recurrence risk in this particular patient cohort.
Collapse
Affiliation(s)
- Dong Xu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Nan Lv
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Qianqian Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Kai Zhang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Jishu Wei
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China.
| | - Min Tu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China.
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Provincial People's Hospital), Pancreas Research Institute, Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China.
| |
Collapse
|
23
|
Lin Z, Rasinski P, Nilsson T, Holstensson M, Song Y, Blomgren A, Jutidamrongphan W, Pandya K, Hong J, Rominger A, Shi K, Axelsson R, Lan X, Seifert R. FAPI PET Versus FDG PET/CT in Gastrointestinal Cancers: An Overview. Semin Nucl Med 2025:S0001-2998(25)00056-X. [PMID: 40399164 DOI: 10.1053/j.semnuclmed.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025]
Abstract
Fibroblast activation protein (FAP) is a type II transmembrane serine protease that is highly expressed in cancer-associated fibroblasts (CAFs) but absent in quiescent fibroblasts. Its overexpression is associated with poor prognosis in various cancers and contributes to treatment resistance. In recent years, radiolabeled FAP inhibitors (FAPI) for PET imaging have shown promising clinical value across a range of cancers. Gastrointestinal (GI) malignancies, which often exhibit a desmoplastic reaction with a high density of FAP-expressing CAFs, are particularly well-suited for FAPI PET. Given the limitations of [18F]FDG PET in GI cancers, such as low sensitivity in certain histological subtypes and high physiological background uptake, FAPI PET is expected to serve as a complementary method, potentially enhancing both diagnostic accuracy and treatment guidance. This review provides a comprehensive comparison of the clinical applications of FAPI PET and [18F]FDG PET in various GI cancers, including their value in diagnosis, staging, and treatment guidance. Additionally, this review summarizes studies on the expanding role of FAPI PET, including its use in assessing treatment response and predicting prognosis, aiming to provide insights into its potential contribution to the improved management of GI malignancies.
Collapse
Affiliation(s)
- Zhaoguo Lin
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Pawel Rasinski
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ted Nilsson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Holstensson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China; Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - August Blomgren
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Warissara Jutidamrongphan
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kalyani Pandya
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jimin Hong
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Rimma Axelsson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China; Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Robert J, Chauhan DS, Cherifi K, Phan QT, Sangwan V, De Crescenzo G, Banquy X. Hybrid Nanocarrier Delivers Immuno-Photothermal Therapy to Modulate Pancreatic Tumor Microenvironment. ACS APPLIED BIO MATERIALS 2025. [PMID: 40390390 DOI: 10.1021/acsabm.5c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers due to its complex tumor microenvironment and limited treatment options. The present study explores a therapeutic strategy that combines immuno- and photothermal therapies (PTT) using hybrid polymer-metal nanoparticles (NPs) to modulate the pancreatic tumor microenvironment, leading to sustained therapeutic efficacy. Core-satellite particles constituted of gold nanorods grafted at the surface of polylactic polyethylene block copolymer particles were designed to encapsulate a potent PI3K-γ inhibitor. The release of the drug from the particles was controlled by near-infrared laser irradiation power and time, offering versatility in controlling dosage profile noninvasively over 96 h. In vitro, 2D cultures of pancreatic cancer cells (KPC) exhibited significantly higher uptake of the hybrid nanoparticles compared to proinflammatory (M1) and anti-inflammatory (M2) macrophages. Consequently, KPC cells were more sensitive to PTT and could be eradicated while maintaining macrophages' viability. Through the photostimulated release of the PI3K-γ inhibitor, the particles effectively repolarized M2 macrophages to the M1 phenotype, enhancing cancer cell eradication. These positive outcomes were further confirmed on 3D cocultures of KPC and macrophage spheroids. Additionally, we showed that macrophages exposed to the nanoparticles exhibited sustained antitumor activity when repeatedly put in contact with cancer cells, confirming the long-term efficacy of the treatment. This study highlights the potential of the present polymer-metal hybrid nanoparticles as a versatile platform for combined immuno- and photothermal therapy in PDAC.
Collapse
Affiliation(s)
- Jordan Robert
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
| | - Deepak S Chauhan
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
| | - Katia Cherifi
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
| | - Quoc Thang Phan
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
| | - Veena Sangwan
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, Québec H4A 0B1, Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal, 2500 Chemin de Polytechnique, Montréal, Québec H3C 3A7, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
- Biomedical Engineering Institute, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
- Chemistry Department, Faculty of Arts and Sciences, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
25
|
Liu Y, Ran X, Zhou G, Liu Y, Tan W. Multivalent Aptamer Assembly Enhances Tumor-Specific Degradation of Transforming Growth Factor-Beta to Remodel the Stromal and Immunosuppressive Cancer Microenvironment. ACS NANO 2025; 19:18164-18175. [PMID: 40326636 DOI: 10.1021/acsnano.4c16628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Extracellular proteins like transforming growth factor-β (TGFβ) are crucial enforcers in the development of cancer stroma and the tumor immunosuppressive microenvironment. Lysosome-targeting chimera-mediated protein degradation appeared as a promising tool for extracellular signal interference but was limited by several lysosome-trafficking receptors and inadequate in vivo degradation efficiency. Here, we designed a multivalent aptamer assembly with a universal pattern to drag extracellular proteins (e.g., TGFβ1) for lysosome degradation with high tumor specificity. By accelerating cell recognition-internalization and lysosomal delivery, the assembly promoted TGFβ blockade and degradation in pancreatic cancer cells and pancreatic stellate cells (PSCs). In vivo, the assembly exhibited highly tumor-specific accumulation and prolonged retention, which resulted in efficient TGFβ inhibition, stromal remodeling, and reversed polarization of immunosuppressive cells in the tumor microenvironment, as well as synergic therapeutic effects when combined with gemcitabine or ovalbumin. Therefore, this study provides a feasible strategy to construct a multivalent aptamer assembly for tumor-specific extracellular protein degradation, after remodeling the tumor stromal and immunosuppressive microenvironment in a manner that enhances the effects of cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinyue Ran
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guangdong Zhou
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
26
|
He R, Shen Z, Chen Q, Hu H, Ding X, Zheng Z, Feng Q, Li B. Pancreatic cancer mortality in China from 2004 to 2021: an in-depth analysis of age, gender, and regional disparities. BMC Cancer 2025; 25:891. [PMID: 40389880 PMCID: PMC12087137 DOI: 10.1186/s12885-025-13863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE This study aimed to analyze the trends and epidemiological characteristics of pancreatic cancer (PC) mortality in China from 2004 to 2021, focusing on gender, age, and regional disparities. The goal was to provide a comprehensive understanding of PC mortality and identify key risk factors to support future prevention and control strategies. METHODS Using data from the national Disease Surveillance Point (DSP) system, which covers a large and representative sample of the Chinese population, the study examined pancreatic cancer mortality trends across different age groups, sexes, and regions. Statistical analyses, including the independent-sample t-test and age-period-cohort (APC) model, were employed to assess mortality differences and annual percentage changes from 2004 to 2021. RESULTS The study recorded a significant increase in pancreatic cancer mortality, particularly among males and older adults. Mortality was consistently higher in urban areas, but the growth rate in rural areas surpassed that of urban areas. Regional disparities were also observed, with the eastern region showing the highest mortality rates but slower increases compared to the central and western regions. Key risk factors, including aging, diabetes, smoking, and chronic pancreatitis, were identified, with gender-specific differences linked to lifestyle factors such as smoking and alcohol consumption. CONCLUSION Pancreatic cancer mortality in China has shown significant increases over the past 18 years, especially among males, older adults, and rural populations. The findings highlight the urgent need for targeted public health interventions to address gender- and age-specific risks, as well as healthcare access inequalities in less developed regions. Future research should focus on gathering more granular, individual-level data to better understand the complex interplay of risk factors and inform more effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Rui He
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhengnan Shen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiuping Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Haiyang Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Ding
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhenglong Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Quansheng Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Baixue Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
27
|
Wang P, Guo W, Liu S, Li S, Li J, Ding B, Yin F, Yang Y, Li X, Cao P, Ma C, Zhang W, Song Y, Geng Y, Liu L, Hu J, Hao J, Feng Y. Novel Pt@PCN-Cu-induced cuproptosis amplifies αPD-L1 immunotherapy in pancreatic ductal adenocarcinoma through mitochondrial HK2-mediated PD-L1 upregulation. J Exp Clin Cancer Res 2025; 44:149. [PMID: 40382627 DOI: 10.1186/s13046-025-03409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Copper accumulation triggers mitochondrial-driven cell death, known as cuproptosis, offering a promising mechanism for targeted cancer therapy. Recent studies have highlighted the critical role of intratumoral copper levels in regulating the expression of programmed cell death ligand-1 (PD-L1), suggesting that copper-induced cuproptosis not only enhances cancer cell death but may also amplify the effects of anti-PD-L1 antibodies (αPD-L1). However, in tumors where monotherapy with αPD-L1 shows limited efficacy, particularly in pancreatic ductal adenocarcinoma (PDAC), the role of copper-induced cuproptosis in enhancing αPD-L1 treatment efficacy and its underlying mechanisms remain unclear. Meanwhile, inadequate tumor drug accumulation and glycolysis significantly restrict the efficacy of cuproptosis. To address these challenges, we have synthesized a novel nanozyme, Pt@PCN-Cu, designed to stabilize intracellular copper accumulation and effectively induce cuproptosis. Additionally, we aim to determine whether this strong induction of cuproptosis can synergize with αPD-L1 to enhance cancer therapy, ultimately paving the way for novel strategies to improve PDAC treatment. METHODS Pt@PCN-Cu was synthesized via a one-pot method, and its therapeutic potential was assessed in combination with αPD-L1 for the treatment of PDAC. Initially, the material's properties were characterized, and its efficient cellular uptake was confirmed. Anti-tumor efficacy was evaluated by inducing cuproptosis in PDAC cell lines and xenograft models. RNA sequencing (RNA-seq) was utilized to identify key regulators involved in the modulation of PD-L1 expression by cuproptosis. Lastly, the therapeutic efficacy of Pt@PCN-Cu combined with αPD-L1 was evaluated in vivo, focusing on tumor growth inhibition and immune modulation within the tumor microenvironment (TME). RESULTS Pt@PCN-Cu demonstrates excellent physicochemical properties and remarkable cascade catalytic activity, providing a solid foundation for further in vitro and in vivo studies. In vitro, Pt@PCN-Cu efficiently transports copper and induces cuproptosis primarily through mitochondrial dysfunction. Mechanistic studies show that Pt@PCN-Cu triggers the dissociation of hexokinase 2 (HK2) from mitochondria, leading to a reduction in HK2 activity. This decline in HK2 activity impairs glycolysis, a metabolic pathway essential for tumor energy metabolism, which in turn results in elevated PD-L1 levels. In vivo, Pt@PCN-Cu demonstrates excellent safety and accumulates at the tumor site in a subcutaneous PDAC mouse model, inducing cuproptosis. Moreover, the combination of Pt@PCN-Cu with αPD-L1 further enhanced its therapeutic efficacy and effectively reprogrammed the immunosuppressive TME. CONCLUSION This study presents strong evidence confirming the safety and therapeutic potential of Pt@PCN-Cu in PDAC treatment. Importantly, Pt@PCN-Cu not only induces cuproptosis but also significantly enhances antitumor efficacy in combination with αPD-L1 by regulating PD-L1 expression through HK2 modulation. These findings underscore a more effective and innovative approach for treating PDAC.
Collapse
Affiliation(s)
- Pengyu Wang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Weihua Guo
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shuyue Liu
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shouyi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiaqi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Bowen Ding
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Fengyi Yin
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yang Yang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xingjiang Li
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Pei Cao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chaozhe Ma
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Wanying Zhang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yating Geng
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China.
| | - Jing Hu
- School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, China.
| | - Jihui Hao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Yukuan Feng
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
28
|
Jiang Y, Ji D, Chen W, Zhu Y, Luo M, Zou R, Fu Y, Huang P, Shi Q, Wang D, Song Z. Phosphorylation of USP32 by CDK5 regulates Rap1 stability and therapeutic resistance in pancreatic ductal adenocarcinoma. Oncogene 2025:10.1038/s41388-024-03263-2. [PMID: 40379759 DOI: 10.1038/s41388-024-03263-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 05/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal human cancer. Gemcitabine-based chemotherapy remains the cornerstone for advanced PDAC. However, resistance to chemotherapy greatly limits its clinical therapeutic efficacy. Accordingly, the identification of novel therapeutic targets to overcome chemoresistance and improve prognosis is urgently needed. Screening of deubiquitinase family members, tandem affinity purification, mass spectrometry, and RNA sequencing (RNA-Seq) analysis were performed to predict the interactions and function of the CDK5-USP32-Rap1 axis in PDAC. In vitro and in vivo experiments were performed to elucidate the regulatory mechanism and biological roles of this axis in glycolytic reprogramming and chemoresistance in PDAC. Finally, TCGA database analysis and immunohistochemistry were performed to determine the expression and clinical significance of CDK5, USP32, and Rap1 in PDAC tissues. USP32 was identified as a bona fide deubiquitinase of Rap1. USP32 deubiquitinates and stabilizes Rap1, thereby promoting glycolytic reprogramming and chemoresistance in PDAC cells. Moreover, we unexpectedly found that CDK5-mediated phosphorylation of USP32 is required for its deubiquitinase activity toward Rap1 and drives malignant phenotypes of PDAC. Additionally, these functions can be significantly inhibited by pharmacological inhibition (roscovitine) or genetic ablation of CDK5. Importantly, combining a CDK5 inhibitor with gemcitabine has a synergetic anticancer effect. Indeed, the effectiveness of targeting CDK5 to sensitize PDAC cells to gemcitabine was confirmed in a patient-derived xenograft (PDX) model. CDK5 and USP32 expression is markedly elevated in PDAC samples and positively associated with Rap1 expression. Increased expression of CDK5, USP32, and Rap1 is significantly associated with poorer prognosis in PDAC. We identified the previously unrecognized oncogenic function and clinical importance of the CDK5-USP32-Rap1 axis, providing preclinical evidence for potential new combination strategies for PDAC therapy.
Collapse
Affiliation(s)
- Yanxia Jiang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dexiang Ji
- Department of Hematology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wen Chen
- Department of Breast Surgery, Jiangxi Cancer Hospital, the Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, 330029, Jiangxi, China
| | - Yuanzhe Zhu
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ming Luo
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Rui Zou
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yilun Fu
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ping Huang
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qing Shi
- Department of Endocrinology and Metabolism, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Dejie Wang
- Department of Gastroenterology, Collaborative Innovation Center of Gastroenterology, Angiocardiopathy and Neurosciences, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| | - Zhiwang Song
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
29
|
Seiki Y, Ikezawa K, Kai Y, Takada R, Kawabata M, Kishimoto H, Hosokawa K, Watsuji K, Kozumi K, Urabe M, Mukai K, Nakabori T, Ohkawa K. Impact of zinc deficiency on the prognosis of unresectable pancreatic cancer. Pancreatology 2025:S1424-3903(25)00092-4. [PMID: 40410049 DOI: 10.1016/j.pan.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/28/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer (PC) is an aggressive malignancy with a poor prognosis. Zinc deficiency (ZD) is prevalent among patients with cancer, especially those with PC, owing to factors such as malabsorption and systemic inflammation. Although ZD is associated with poor nutritional status and cancer cachexia, its impact on overall survival (OS) in unresectable PCs remains unclear. This study aimed to evaluate the prevalence of ZD in patients with unresectable PC and its influence on prognosis. METHODS A retrospective study was conducted on 272 patients with unresectable pancreatic cancer whose serum zinc levels measured before starting chemotherapy between April 2016 and March 2024. ZD was defined as serum zinc level below 60 μg/dL. Patient characteristics, nutritional indices (modified Glasgow prognostic score [mGPS], neutrophil-to-lymphocyte ratio [NLR], prognostic nutritional index [PNI], controlling nutritional status [CONUT] score), and survival outcomes were analyzed. RESULTS ZD was identified in 19.4 % of patients. Patients with ZD were significantly older and exhibited lower PNI, indicating poorer nutritional status. Regarding OS, univariate analysis showed that the ZD group had significantly lower survival rates than non-ZD group. Multivariate analysis revealed that ZD was an independent predictor of poor prognosis. CONCLUSIONS This study demonstrates that ZD before chemotherapy is an independent prognostic factor in patients with unresectable PC undergoing chemotherapy. Considering its association with poor nutritional status, serum zinc levels may serve as a valuable prognostic biomarker. Further research is warranted to explore the potential therapeutic benefits of zinc supplementation therapy.
Collapse
Affiliation(s)
- Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan.
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masaki Kawabata
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroki Kishimoto
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kana Hosokawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuhiro Kozumi
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
30
|
Zhang T, Celiker B, Shao Y, Gai J, Hill M, Wang C, Zheng L. Comparison of Shared Class I HLA-Bound Noncanonical Neoepitopes between Normal and Neoplastic Tissues of Pancreatic Adenocarcinoma. Clin Cancer Res 2025; 31:1956-1965. [PMID: 39699517 PMCID: PMC12079097 DOI: 10.1158/1078-0432.ccr-24-2251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE Developing T-cell or vaccine therapies for pancreatic ductal adenocarcinoma (PDAC) has been challenging because of a lack of knowledge regarding immunodominant, cancer-specific antigens as PDAC are characterized by a scarcity of genomic mutation-associated neoepitopes, and effective approaches to discover them are limited. EXPERIMENTAL DESIGN An advanced mass spectrometry approach was employed to compare the immunopeptidome of PDAC tissues and matched normal tissues from the same patients. RESULTS This study identified HLA class I-binding variant peptides derived from canonical proteins, which had single amino-acid substitutions not attributed to genetic mutations or RNA editing. These amino-acid substitutions appeared to result from translational errors. The variant peptides were predominantly found in tumor tissues, with certain peptides common among multiple patients. Importantly, several of these variant peptides were more immunogenic than their wild-type counterparts. CONCLUSIONS The shared noncanonical neoepitopes identified in this study offer promising candidates for vaccine and T-cell therapy development, potentially providing new avenues for immunotherapy in PDAC. See related commentary by Yuan et al., p. 1821.
Collapse
Affiliation(s)
- Tengyi Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Betul Celiker
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yingkuan Shao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Breast Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Cancer Institute, Ministry of Education, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jessica Gai
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Hill
- Immuno-Oncology Discovery and Translational Medicine, Bristol Myers Squibb Company, Seattle, Washington
| | - Chunyu Wang
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Cancer Convergence Institute at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Yang X, Liang N, Liu D, Yan J, Yang X, Lv J, Xiao S, Wei X, Chen X, Yang Z, Gui S, Jin L, Yu S, Lyu J, Ren X. Antioxidant capacity of the iron-sulfur cluster assembly protein IscU2 is mediated by aspartate metabolism to promote tumor survival. J Biol Chem 2025:110234. [PMID: 40378953 DOI: 10.1016/j.jbc.2025.110234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Environmental nutrient levels affect cancer cell metabolism, activating adaptive mechanisms in cancer cells to deal with nutrient stress. However, it remains unclear how tumor cells sustain survival under nutrient-stress circumstances through metabolic reprogramming. Our study focused on nutrient deficiency-induced oxidative damage, revealing that increased expression of the iron-sulfur (Fe-S) cluster assembly protein, IscU2, is essential for the survival of pancreatic ductal adenocarcinoma (PDAC) cells in glucose-deficient conditions. Glucose deficiency induces IscU2 expression via the activation of the AMPK pathway, allowing IscU2 to exhibit antioxidant properties that are absent under glucose-sufficient conditions. Upregulated IscU2 stimulates aspartate synthesis by bolstering mitochondrial metabolism, including respiration and the tricarboxylic acid cycle, in a Fe-S cluster-dependent manner. Notably, oxidative stress and apoptosis induced by IscU2 depletion in glucose-deficient PDAC cells can be restored by aspartate-mediated NADPH production. These findings highlight the importance of IscU2 in PDAC cell metabolism and its essential function in supporting cell survival under nutrient-deficient conditions.
Collapse
Affiliation(s)
- Xunjun Yang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Laboratory Medicine, Wenzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Liang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dandan Liu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jimei Yan
- Department of Laboratory Medicine, Linyi Peoples' Hospital, Linyi, Shandong, China
| | - Xiali Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinya Lv
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Saijun Xiao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiujuan Wei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xuyang Chen
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhengquan Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shanying Gui
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Liqin Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shihui Yu
- School of Basic Medical Sciences, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Jianxin Lyu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Xiaojun Ren
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
32
|
Girolimetti G, Gagliardi S, Cordella P, Bramato G, Di Corato R, Romano R, Guerra F, Bucci C. Induced mitochondrial deficit by NDUFS3 transient silencing reduces RAB7 expression and causes lysosomal dysfunction in pancreatic cancer cells. Cell Commun Signal 2025; 23:224. [PMID: 40369571 PMCID: PMC12079996 DOI: 10.1186/s12964-025-02214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND RAB7 is a small GTPase with multiple cellular roles, regulating late endocytic trafficking and lysosomal biogenesis, influencing mitochondria-lysosome crosstalk, and contributing to many mitochondrial processes. Mitochondrial dysfunctions are widely reported in cancer and the development of cancer therapeutic strategies targeting mitochondria gained momentum in recent years. Mitochondrial impairment can cause alterations of mitochondria-lysosome crosstalk and can influence lysosomal function. Here, we used cell models of pancreatic cancer, one of the deadliest cancers worldwide, to cause a transient mild mitochondrial deficit lowering NDUFS3 protein levels in order to investigate the consequences on RAB7 and on the late endocytic pathway and, thus, the contribution of the mitochondria-lysosomes communication alterations to cancer progression. METHODS NDUFS3 and RAB7 downregulation was obtained by RNA interference (RNAi). Seahorse assays, Western blot analysis, mitochondrial staining, and Transmission Electron Microscopy (TEM) were used to assess silencing effects on mitochondrial structure and functioning. Western blotting was used to investigate expression of late endocytic pathway proteins and of the invasion marker vimentin. Confocal microscopy was used to analyze the mitochondrial network and lysosomal assessment. Zymography was performed to evaluate the ability to digest the extracellular matrix linked to cancer migration. SRB and colony assays were performed to assess cancer viability and proliferation. Wound healing assay and FluoroBlok membranes were used to determine migration and invasiveness. RESULTS In pancreatic cancer cells, transient silencing of the NDUFS3 protein caused mitochondrial deficit, slower oxidative metabolism, and mitochondrial morphology alterations. In this context, we observed RAB7 downregulation and impairment of the late endocytic pathway. In addition, NDUFS3-silenced RAB7-downregulated cells showed less invasive tumorigenic potential revealed by reduced levels of vimentin and other Epithelial-to-Mesenchymal Transition proteins, decreased viability, migration and invasiveness. Moreover, we found that modulation of RAB7 expression may regulate vimentin levels and influence mitochondrial morphology and levels of mitochondrial proteins. CONCLUSIONS Overall, our data show that mitochondrial deficit determines alterations of the crosstalk with lysosomes, leading to dysfunctions, and that this process is regulated by RAB7 acting as an oncogene. This highlights the synergic role of RAB7 and mitochondrial dysfunction, focusing on a cellular mechanism that may boost the effect of mitochondrial dysfunction in the cells, leading to the reduction of the tumorigenic potential.
Collapse
Affiliation(s)
- Giulia Girolimetti
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Sinforosa Gagliardi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Paola Cordella
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Grazia Bramato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Riccardo Di Corato
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Provinciale Lecce-Monteroni, Lecce, 73100, Italy
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, Arnesano, 73010, Italy
| | - Roberta Romano
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy.
| |
Collapse
|
33
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
34
|
Hu X, Wang Z, Zhu Y, Li Z, Yan H, Zhao X, Wang Q. Advancements in molecular imaging for the diagnosis and treatment of pancreatic ductal adenocarcinoma. NANOSCALE ADVANCES 2025; 7:2887-2903. [PMID: 40270837 PMCID: PMC12012634 DOI: 10.1039/d4na01080a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor characterized by poor overall patient survival and prognosis, largely due to challenges in early diagnosis, limited surgical options, and a high propensity for therapy resistance. The integration of various imaging modalities through molecular imaging techniques, particularly multimodal molecular imaging, offers the potential to provide more precise and comprehensive information about the lesion. With advances in nanomedicine, new imaging and drug delivery approaches that allow the development of multifunctional theranostic agents offer opportunities for improving pancreatic cancer treatment using precision oncology. Herein, we review the diagnostic and therapeutic applications of molecular imaging for PDAC and discuss the adoption of multimodal imaging approaches that combine the strengths of different imaging techniques to enhance diagnostic accuracy and therapeutic efficacy. We emphasize the significant role of nanomedicine technology in advancing multimodal molecular imaging and theranostics, and their potential impact on PDAC management. This comprehensive review aims to serve as a valuable reference for researchers and clinicians, offering insights into the current state of molecular imaging in PDAC and outlining future directions for improving early diagnosis, combination therapies, and prognostic evaluations.
Collapse
Affiliation(s)
- Xun Hu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zihua Wang
- School of Basic Medical Sciences, Fujian Medical University Fuzhou 350122 Fujian Province China
| | - Yuting Zhu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital Shenzhen Guangdong 518036 China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School/Tsinghua University Shenzhen 518055 China
| | - Xinming Zhao
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| |
Collapse
|
35
|
Jia Y, Wang Y, Zhao G, Yang Y, Yan W, Wang R, Han B, Wang L, Zhang Z, Chen L, Lemoine NR, Chard Dunmall LS, Wang P, Wang Y. Novel oncolytic vaccinia virus armed with interleukin-27 is a potential therapeutic agent for the treatment of murine pancreatic cancer. J Immunother Cancer 2025; 13:e010341. [PMID: 40350204 PMCID: PMC12067774 DOI: 10.1136/jitc-2024-010341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Pancreatic cancer has a complex immunosuppressive tumor microenvironment (TME), which is highly resistant to conventional therapies and emerging cancer immunotherapies. Oncolytic viruses are multifaceted killers of malignant tumors, which can selectively infect, replicate in and lyse tumor cells, release tumor-associated antigens to stimulate specific antitumor immune responses, and recruit immune cells into the TME, turning "cold" tumors "hot". Here, we report a novel vaccinia virus (VV), VVLΔTKΔN1LΔA41L (with deletion of thymidine kinase (TK), N1L, and A41L genes) armed with interleukin 27 (IL-27), that can cure established tumors and promote long-term antitumor immunity in murine pancreatic cancer tumor models. METHODS A novel oncolytic VV with deletion of the TK, N1L, and A41L genes, and expression of the red fluorescent protein (RFP) gene (VVL-TD-RFP) was constructed using CRISPR-Cas9-based homologous recombination. This virus was armed with IL-27, creating VVL-TD-IL-27. The characteristics of these viruses were evaluated in vitro using viral replication assays, cytotoxicity assays and ELISA. The antitumor effects of VVL-TD-IL-27 were evaluated using a variety of pancreatic cancer tumor models in vivo, and the mechanisms of antitumor effects were explored using flow cytometry, immunohistochemistry, ELISA and quantitative PCR. RESULTS VVL-TD-RFP cured 71.4% of tumor-bearing mice, compared with 14.3% of animals treated with VVLΔTKΔN1L that does not have an A41L gene deletion. Efficacy was mainly dependent on elevated dendritic cell (DC) populations, activation of DC, CD86+ DC, and CD8+ effector memory T cells in the TME. Efficacy was further enhanced by arming VVL-TD-RFP with IL-27, which resulted in a cure rate of 100% and promoted long-term antitumor immunity. VVL-TD-IL-27 treatment increased the proportion of CD8+ TEM and decreased the proportion of regulatory T cells and macrophages in tumor tissues. It also polarized macrophages to an M1 phenotype in vivo. Furthermore, IL-27 exhibits strong anti-angiogenic effects. CONCLUSIONS VVL-TD-mIL-27 is a potential immunotherapy agent for the treatment of pancreatic cancer, and a clinical study of this virus is warranted.
Collapse
Affiliation(s)
- Yangyang Jia
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanru Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Guanghao Zhao
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yong Yang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenyi Yan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ruimin Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bing Han
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lihong Wang
- Department of Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Zhe Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijuan Chen
- Department of Oncology, Henan International Joint Laboratory of Lung Cancer Biology and Therapeutics, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Nicholas R Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
36
|
Li Z, Zhang J, Yin J, Ma W, Liao H, Ling L, Zou Q, Cao Y, Song Y, Zheng G, Hu X, Yang G, Li N. Targeting MYOF suppresses pancreatic ductal adenocarcinoma progression by inhibiting ILF3-LCN2 signaling through disrupting OTUB1-mediated deubiquitination of ILF3. Redox Biol 2025; 84:103665. [PMID: 40381229 PMCID: PMC12145718 DOI: 10.1016/j.redox.2025.103665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is still a highly aggressive and fatal disease. The molecular mechanisms for PDAC progression are still not fully understood. Here, we demonstrated the overexpression of MYOF in PDAC in multiple sample sets, which is significantly associated with poor outcome of PDAC patients. MYOF knockout suppresses PDAC progression in vitro and in vivo. MYOF knockout exerts its effects by promoting ferroptosis via downregulating LCN2 expression. Ectopic LCN2 expression overcame the effects of MYOF knockout in PDAC cells. Mechanistically, MYOF respectively recruits OTUB1 and ILF3 to enhance their interaction and relieves ILF3 protein ubiquitination and degradtion. MYOF maintains ILF3 protein stability, thereby enhances ILF3 interacting with and improving LCN2 mRNA stability. Moreover, we screened and identified natural compound Picroside II potentially targets MYOF to suppress PDAC progression. These findings uncover the biological roles and mechanisms of MYOF and preliminarily indicate the potential of targeting MYOF in PDAC progression, highlighting a novel therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Zhihui Li
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Jianlei Zhang
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Jiang Yin
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Wen Ma
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Hongfan Liao
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Lv Ling
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Qingfeng Zou
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Yabing Cao
- Kiang Wu Hospital, Macao Special Administrative Region of China
| | - Ying Song
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Guopei Zheng
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China
| | - Xiaoye Hu
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China.
| | - Guohua Yang
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China.
| | - Nan Li
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University; Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Hengzhigang Road 78#, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
37
|
Shen Q, Liu J, Zeng L, Ren Y, Liao J, Chen S, Tang Y, Zhang Z, Jiang M, Liao H, Wang L, Xu X, Chen J. Pancreas-targeted lipid nanoparticles for relatively non-invasive interleukin-12 mRNA therapy in orthotopic pancreatic ductal adenocarcinoma. J Control Release 2025; 381:113588. [PMID: 40032009 DOI: 10.1016/j.jconrel.2025.113588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90 % of pancreatic cancers and shows limited response to immune therapy owing to the highly immunosuppressive tumor microenvironment (TME). Cytokine-encoded mRNA therapy demonstrates a great promise in converting "cold" tumors into "hot" ones, while it is typically administered through intratumoral injection and applicable only to superficial tumors, which limites their application in PDAC. In this study, we design and develop a lipid nanoparticle (LNP) delivery system capable of targeting pancreatic tissue via intraperitoneal (I.P.) injection. This system not only efficiently delivers mRNA to pancreatic tissues but also selectively targets immune cells in PDAC. A single I.P. injection of LNP encapsulating interleukin-12 (IL-12) mRNA (LNP/mIL-12) activates both myeloid and lymphoid cells in PDAC, reprogramming the immunosuppressive TME. Remarkably, I.P. injection of LNP/mIL-12 induces eradication of orthotopic PDAC in some cases. Our work represents the first relatively non-invasive method to deliver IL-12 mRNA for targeted treatment of orthotopic PDAC, offering a novel approach for PDAC immunotherapy.
Collapse
Affiliation(s)
- Qian Shen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Jia Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China; Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ling Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Yupeng Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering 2 Taoyuan Street, Xiangtan 411201, PR China
| | - Jing Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Sijie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Yingsen Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Zixi Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Meng Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Hangping Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering 2 Taoyuan Street, Xiangtan 411201, PR China
| | - Lingyun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China.
| | - Jinjin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China.
| |
Collapse
|
38
|
Mao Y, Li Y, Zheng Z, Xu Y, Ke M, He A, Liang F, Zhang K, Wang X, Gao W, Tian R. All-at-once spatial proteome profiling of complex tissue context with single-cell-type resolution by proximity proteomics. Cell Syst 2025:101291. [PMID: 40345200 DOI: 10.1016/j.cels.2025.101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/01/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Spatial proteomics enables in-depth mapping of tissue architectures, mostly achieved by laser microdissection-mass spectrometry (LMD-MS) and antibody-based imaging. However, trade-offs among sampling precision, throughput, and proteome coverage still limit the applicability of these strategies. Here, we propose proximity labeling for spatial proteomics (PSPro) by combining precise antibody-targeted biotinylation and efficient affinity purification for all-at-once cell-type proteome capture with sub-micrometer resolution from single tissue slice. With fine-tuned labeling parameters, PSPro shows reliable performance in benchmarking against flow cytometry- and LMD-based proteomic workflows. We apply PSPro to tumor and spleen slices, enriching thousands of proteins containing known markers from ten cell types. We further incorporate LMD into PSPro to facilitate comparison of cell subpopulations from the same tissue slice, revealing spatial proteome heterogeneity of cancer cells and immune cells in pancreatic tumor. Collectively, PSPro converts the traditional "antibody-epitope" paradigm to an "antibody-cell-type proteome" for spatial biology in a user-friendly manner. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yiheng Mao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuan Li
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhendong Zheng
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yanfen Xu
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mi Ke
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - An He
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Fuchao Liang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Keren Zhang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Wang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weina Gao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruijun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
39
|
Cao M, Peng W, Cheng B, Wang R, Chen W, Liu L, Huang H, Chen S, Cui H, Liang J, Zhou Q, Xiong S, Bai S, Liu L, Zhao Y. PPY-Induced iCAFs Cultivate an Immunosuppressive Microenvironment in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413432. [PMID: 40162859 PMCID: PMC12120788 DOI: 10.1002/advs.202413432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/20/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by cancer cells surrounded by affluent stromal components, which may underlie their limited response to various therapeutic interventions, including immunotherapy. Inflammatory cancer-associated fibroblasts (iCAFs), a crucial subset of CAFs within the PDAC microenvironment, play a pivotal role in shaping an immunosuppressive microenvironment. In this study, single-cell RNA sequencing analysis is performed to screen for cancer cells-secreted proteins associated with iCAF induction, and PPY (pancreatic polypeptide) is validated as a potent inducer. Unlike previously reported iCAF inducers, PPY is a gastrointestinal hormone predominantly expressed in the pancreas, suggesting that targeting it may have minimal systemic effects. Multiplex immunohistochemistry (mIHC) on human PDAC tissue microarrays, orthotopic allograft mouse models, and co-culture experiments are utilized to validate the crucial role of PPY in iCAF induction. Mechanistic studies integrating mRNA sequencing, immunoprecipitation-mass spectrometry, and molecular docking reveal that PPY induces iCAFs by activating the non-canonical NF-κB pathway through EGFR. Importantly, targeting PPY enhanced the efficacy of anti-PD-1 immunotherapy in KPC (KrasLSL-G12D/+; Trp53LSL-R172H/+; Pdx1-Cre) mice, as evidenced by reduced tumor burden on PET-CT imaging and improved survival. This research is expected to provide a novel strategy for improving immunotherapy in PDAC by targeting a key inducer of iCAFs.
Collapse
Affiliation(s)
- Mengdie Cao
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Wang Peng
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Bin Cheng
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ronghua Wang
- Department of SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Wei Chen
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- School of Life SciencesThe Chinese University of Hong KongShatin, New TerritoriesHong Kong999077China
| | - Luyao Liu
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hai Huang
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shiru Chen
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Haochen Cui
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - JingWen Liang
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Qiaodan Zhou
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Si Xiong
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shuya Bai
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Luoxia Liu
- Department of Nuclear MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yuchong Zhao
- Department of Gastroenterology and HepatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
40
|
Allemailem KS, Rahmani AH, almansour NM, Aldakheel FM, Albalawi GM, Albalawi GM, Khan AA. Current updates on the structural and functional aspects of the CRISPR/Cas13 system for RNA targeting and editing: A next‑generation tool for cancer management (Review). Int J Oncol 2025; 66:42. [PMID: 40342053 PMCID: PMC12068846 DOI: 10.3892/ijo.2025.5748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
For centuries, a competitive evolutionary race between prokaryotes and related phages or other mobile genetic elements has led to the diversification of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR‑associated sequence (Cas) genome‑editing systems. Among the different CRISPR/Cas systems, the CRISPR/Cas9 system has been widely studied for its precise DNA manipulation; however, due to certain limitations of direct DNA targeting, off‑target effects and delivery challenges, researchers are looking to perform transient knockdown of gene expression by targeting RNA. In this context, the more recently discovered type VI CRISPR/Cas13 system, a programmable single‑subunit RNA‑guided endonuclease system that has the capacity to target and edit any RNA sequence of interest, has emerged as a powerful platform to modulate gene expression outcomes. All the Cas13 effectors known so far possess two distinct ribonuclease activities. Pre‑CRISPR RNA processing is performed by one RNase activity, whereas the two higher eukaryotes and prokaryotes nucleotide‑binding domains provide the other RNase activity required for target RNA degradation. Recent innovative applications of the type VI CRISPR/Cas13 system in nucleic acid detection, viral interference, transcriptome engineering and RNA imaging hold great promise for disease management. This genome editing system can also be employed by the Specific High Sensitivity Enzymatic Reporter Unlocking platform to identify any tumor DNA. The discovery of this system has added a new dimension to targeting, tracking and editing circulating microRNA/RNA/DNA/cancer proteins for the management of cancer. However, there is still a lack of thorough understanding of the mechanisms underlying some of their functions. The present review summarizes the recent updates on the type VI CRISPR/Cas system in terms of its structural and mechanistic properties and some novel applications of this genome‑editing tool in cancer management. However, some issues, such as collateral degradation of bystander RNA, impose major limitations on its in vivo application. Furthermore, additional challenges and future prospects for this genome editing system are described in the present review.
Collapse
Affiliation(s)
- Khaled s. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Nahlah Makki almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M. Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Ghadah Mohammad Albalawi
- Department of Laboratory and Blood Bank, King Fahd Specialist Hospital, Tabuk 47717, Saudi Arabia
| | | | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
41
|
Shakiba M, Tuveson DA. Macrophages and fibroblasts as regulators of the immune response in pancreatic cancer. Nat Immunol 2025; 26:678-691. [PMID: 40263612 DOI: 10.1038/s41590-025-02134-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the few cancers that has yet to benefit from immunotherapies. This is primarily a result of its characteristic 'cold' tumor microenvironment composed of cancer-associated fibroblasts (CAFs), a dense network of extracellular matrix and several immune cell types, the most abundant of which are the tumor-associated macrophages (TAMs). Advances in single-cell and spatial technologies have elucidated the vast functional heterogeneity of CAFs and TAMs, their symbiotic relationship and their cooperative role in the tumor microenvironment. In this Review, we provide an overview of the heterogeneity of CAFs and TAMs, how they establish an immunosuppressive microenvironment and their collaboration in the remodeling of the extracellular matrix. Finally, we examine why the impact of immunotherapy in PDAC has been limited and how a detailed molecular and spatial understanding of the combined role of CAFs and TAMs is paramount to the design of effective therapies.
Collapse
Affiliation(s)
- Mojdeh Shakiba
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
42
|
Zhang ZL, Xu HN, Gong CM, Li YZ, Song XM, Li YM, Zhang DD, Wang R. Microorganism-Derived Bisindole Alkaloids With Anticancer Potential and Their Mechanisms: A Comprehensive Review. Chem Biodivers 2025; 22:e202402398. [PMID: 39714457 DOI: 10.1002/cbdv.202402398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Bisindole alkaloids constitute a significant class of natural compounds distinguished by their characteristic bisindole structure and renowned for their anticancer properties. Over the last six decades, researchers have isolated 425 microorganism-derived bisindole alkaloids (MDBAs). Among them, 187 MDBAs have demonstrated anticancer properties against various in vitro cancer cell lines, primarily by impeding the cell cycle, restraining cell proliferation, and inducing apoptosis and autophagy. These effects are mediated by regulating key targets and signaling pathways such as hypoxia-inducible factor (HIF)-1, MAPK, and phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR. This review provides a comprehensive examination of the sources, chemical diversity, and anticancer properties of these compounds. Furthermore, it summarizes the structure-activity relationship (SAR), druggability, and the mechanisms underlying MDBAs' anticancer effects. Ultimately, this article aims to furnish a thorough overview of the advancements in the investigation of microorganism-derived bisindole alkaloids for their continued development and utilization.
Collapse
Affiliation(s)
- Zi-Long Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Hao-Nan Xu
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Chuan-Ming Gong
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Yu-Ze Li
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Xiao-Mei Song
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Yi-Ming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Dong-Dong Zhang
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Rui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
43
|
Hong WC, Kim M, Kim JH, Kang HW, Fang S, Jung HS, Kwon W, Jang JY, Kim HJ, Park JS. The FOXP1-ABCG2 axis promotes the proliferation of cancer stem cells and induces chemoresistance in pancreatic cancer. Cancer Gene Ther 2025; 32:563-572. [PMID: 40169859 PMCID: PMC12086089 DOI: 10.1038/s41417-025-00896-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025]
Abstract
Pancreatic cancer is an aggressive disease with low survival and high recurrence rates. A major obstacle in treating pancreatic cancer is the frequent development of chemoresistance to the standard therapeutic drug, gemcitabine. One mechanism by which pancreatic cancer develops chemoresistance is through the proliferation of cancer stem cells (CSC). However, the mechanisms regulating stemness in chemoresistant tumors remain unclear. Here, we found that the expression of the transcription factor Forkhead Box P1 (FOXP1) was elevated in chemoresistant pancreatic cancer and crucial for establishing CSC characteristics. Silencing FOXP1 reduced the expressions of stemness-associated genes and diminished the formation of both spheroids and colonies, highlighting the crucial role of FOXP1 in regulating stemness in chemoresistant tumor cells. Mechanistically, we discovered that FOXP1 regulates the expression of ATP-binding cassette superfamily G member 2 (ABCG2), which induces the efflux of gemcitabine. Knockdown of FOXP1 reduced the expression of ABCG2, resulting in decreased proliferation and increased sensitivity to gemcitabine. Moreover, the inhibition of FOXP1 in orthotopic mouse models reduced tumor growth and proliferation, and enhanced sensitivity to gemcitabine. Together, our data reveal FOXP1 as a potent oncogene that promotes CSC growth in chemoresistant pancreatic cancer.
Collapse
Affiliation(s)
- Woosol Chris Hong
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsoo Kim
- Korea Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Ju Hyun Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeon Woong Kang
- Korea Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Sungsoon Fang
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Figlioli G, Piovani D, Tsantes AG, Pugliese N, Nikolopoulos GK, Hassan C, Repici A, Lleo A, Aghemo A, Bonovas S. Burden of cancer attributable to high body mass index: A systematic analysis of the Global Burden of Disease Study 2021. Clin Nutr 2025; 48:144-152. [PMID: 40215883 DOI: 10.1016/j.clnu.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND High body mass index (BMI) is a well-established cancer risk factor. Reliable, updated data are essential for guiding public health policies and designing effective interventions to reduce the cancer burden associated with high BMI. METHODS Data from the Global Burden of Disease Study 2021 on cancer burden attributable to high BMI were analysed globally, stratified by sex, age, geographic region, cancer type, and socio-demographic index (SDI). Temporal trends in age-standardized rates from 1990 to 2021 were evaluated using estimated annual percentage changes. RESULTS In 2021, cancer attributable to high BMI resulted in 356.74 thousand deaths (95% uncertainty interval: 146.12-581.01) and 8.89 million (3.75-14.38) Disability-Adjusted Life Years (DALYs), with females bearing the largest burden. From 1990 to 2021, age-standardized rates of high BMI-related cancer deaths increased by 0.35% annually, while DALYs rose by 0.42% annually. In 2021, the burden of cancer deaths and DALYs attributable to high BMI varied considerably across geographical regions. Low-middle SDI regions experienced the largest increases in death and DALY rates attributable to high BMI, while these rates declined in high SDI regions. Colon and rectum cancers accounted for the greatest number of deaths and DALYs, while pancreatic cancer showed the most rapid growth in attributable burden. CONCLUSIONS High BMI is a major contributor to the global cancer burden, with significant variation by sex, cancer type, region, and SDI level. Targeted public health strategies are urgently needed to mitigate the growing impact of overweight and obesity on cancer.
Collapse
Affiliation(s)
- Gisella Figlioli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andreas G Tsantes
- Laboratory of Haematology and Blood Bank Unit, "Attiko" Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicola Pugliese
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alessio Aghemo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Stefanos Bonovas
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| |
Collapse
|
45
|
Yin L, Cao C, Lin J, Wang Z, Peng Y, Zhang K, Xu C, Yang R, Zhu D, Wang F, Chang S, Bao H, Yang S, Li N, Wu X, Shao Y, Wu Z, Wu S, Pu N, Xu Z, Guo F, Feng X, Chen J, Xiao B, Tu M, Li Q, Wei J, Wu J, Gao W, Miao Y, Liu L, Lu Z, Jiang K. Development and Validation of a Cell-Free DNA Fragmentomics-Based Model for Early Detection of Pancreatic Cancer. J Clin Oncol 2025:JCO2400287. [PMID: 40311105 DOI: 10.1200/jco.24.00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/08/2025] [Accepted: 03/05/2025] [Indexed: 05/03/2025] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC), known for its high fatality rate, is often diagnosed in its advanced stages where surgical options are not viable. This highlights the critical need for innovative and effective early detection techniques. This study focuses on the potential of cell-free DNA (cfDNA) fragmentomics integrating advanced machine learning to identify early-stage PDAC with high accuracy. METHODS Our study included a broad cohort of 1,167 participants, from which plasma was collected and subjected to shallow whole-genome sequencing. After rigorous quality assessments, 166 individuals diagnosed with PDAC and 167 healthy participants were in the training cohort, whereas the validation cohort consisted of 112 patients with PDAC and 111 healthy individuals. A separate group of 67 individuals with nonmalignant pancreatic cysts was also included to validate the model's accuracy. Finally, two additional external validation cohorts and one additional independent early-stage data set were included to evaluate the robustness of model. Our analysis used fragmentomic profiling, integrating copy-number variations, fragment size, mutational signatures, and methylation patterns analyzed using machine learning. RESULTS The model demonstrated remarkable accuracy in distinguishing patients with PDAC from controls, with an AUC of 0.992 in the training data set and 0.987 in the validation data set. At a cutoff of 0.52, the training set reached a sensitivity of 93.4% and a specificity of 95.2%. In the validation data set, the sensitivity was 97.3% with a specificity of 92.8%, while the external data set demonstrated a sensitivity of 90.91% and a specificity of 94.5%. CONCLUSION This study underscores the effectiveness of using cfDNA fragmentomics and machine learning for early detection of PDAC. Our approach promises significant potential in reducing PDAC mortalities through early intervention and could serve as a breakthrough in oncologic diagnostics.
Collapse
Affiliation(s)
- Lingdi Yin
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Cheng Cao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Jianzhen Lin
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yunpeng Peng
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Kai Zhang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Cheng Xu
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruowei Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Dongqin Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Shuang Chang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Shanshan Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Ningyou Li
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Guo
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Xu Feng
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Jianmin Chen
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Bin Xiao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Min Tu
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Qiang Li
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Jishu Wei
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Junli Wu
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Wentao Gao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zipeng Lu
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute of Nanjing Medical University, Nanjing, China
| |
Collapse
|
46
|
Bos MD, Meyer NH, Wijma AG, Khatib-Chahidi K, van den Broek E, Ho CS, Meerdink M, Klaase JM, Bockhorn M, Hoogwater FJ, Nijkamp MW. Preoperative Anemia as a Prognostic Risk Factor for Inferior Oncological Survival Following Resection for Pancreatic Ductal Adenocarcinoma. Pancreas 2025; 54:e397-e406. [PMID: 39854689 PMCID: PMC12052068 DOI: 10.1097/mpa.0000000000002448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/13/2024] [Indexed: 01/26/2025]
Abstract
OBJECTIVES A significant proportion of patients undergoing surgery for pancreatic ductal adenocarcinoma (PDAC) are anemic at the time of resection. In these patients, blood transfusions are omitted because of their potential negative impact on oncological outcomes. The present study aimed to determine the prognostic value of preoperative anemia in resected PDAC patients, irrespective of blood transfusion status. MATERIALS AND METHODS This retrospective 2-center cohort study included patients who underwent resection for PDAC between 2013 and 2022. The prognostic role of preoperative anemia was investigated using Cox proportional-hazard regression analysis. A subgroup analysis excluded PDAC patients who received a perioperative blood transfusion. RESULTS Among 280 included PDAC patients, 110 (39%) were found to have preoperative anemia. Preoperative anemia was associated with increased use of blood transfusions, with 44 patients (16%) requiring transfusion perioperatively. In the whole cohort, preoperative anemia was an independent predictor of lower disease-free survival (hazard ratio [HR] = 1.518; 95% confidence interval [CI] = 1.103-2.090, P = 0.011), but not overall survival. However, when patients who received a perioperative blood transfusion were excluded, preoperative anemia was independently associated with both lower disease-free survival (HR = 1.636; 95% CI = 1.113-2.404, P = 0.012) and overall survival (HR = 1.484; 95% CI = 1.036-2.127, P = 0.031). CONCLUSIONS Preoperative anemia was identified as an independent risk factor for inferior oncological survival after resection for PDAC. These results support the need for increased awareness regarding the potential adverse effects of preoperative anemia on oncological outcomes in PDAC.
Collapse
Affiliation(s)
- Mylena D. Bos
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - N. Helge Meyer
- Department of Human Medicine, University Hospital of General and Visceral Surgery, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Allard G. Wijma
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Karl Khatib-Chahidi
- Department of Human Medicine, University Hospital of General and Visceral Surgery, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Evert van den Broek
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cassandra S.L. Ho
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Human Medicine, University Hospital of General and Visceral Surgery, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Mark Meerdink
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joost M. Klaase
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maximilian Bockhorn
- Department of Human Medicine, University Hospital of General and Visceral Surgery, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Frederik J.H. Hoogwater
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maarten W. Nijkamp
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
47
|
Shi J, Cui X, Wang J, Liu G, Meng J, Zhang Y. Crosstalk between the tumor immune microenvironment and metabolic reprogramming in pancreatic cancer: new frontiers in immunotherapy. Front Immunol 2025; 16:1564603. [PMID: 40356913 PMCID: PMC12066759 DOI: 10.3389/fimmu.2025.1564603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
In recent years, the incidence and mortality of pancreatic cancer (PC) are increasing year by year. The highly heterogeneous nature of PC, its strong immune escape ability and easy metastasis make it the most lethal malignant tumor in the world. With the rapid development of sequencing technology, the complex components in the tumor microenvironment (TME) of PC have been gradually revealed. Interactions between pancreatic stellate cells, tumor-associated fibroblasts, various types of immune cells, and cancer cells collectively promote metabolic reprogramming of all types of cells. This metabolic reprogramming further enhances the immune escape mechanism of tumor cells and ultimately induces tumor cells to become severely resistant to chemotherapy and immunotherapy. On the one hand, PC cells achieve re and rational utilization of glucose, amino acids and lipids through metabolic reprogramming, which in turn accomplishes biosynthesis and energy metabolism requirements. Under such conditions, tumorigenesis, proliferation and metastasis are ultimately promoted. On the other hand, various types of immune cells in the tumor immune microenvironment (TIME) also undergo metabolic reprogramming, which leads to tumor progression and suppression of anti-immune responses by inhibiting the function of normal anti-tumor immune cells and enhancing the function of immunosuppressive cells. The aim of this review is to explore the interaction between the immune microenvironment and metabolic reprogramming in PC. The focus is to summarize the specific mechanisms of action of metabolic reprogramming of PC cells and metabolic reprogramming of immune cells. In addition, this review will summarize the mechanisms of immunotherapy resistance in PC cells. In the future, targeting specific mechanisms of metabolic reprogramming will provide a solid theoretical basis for the development of combination therapies for PC.
Collapse
Affiliation(s)
- Jintai Shi
- College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoyan Cui
- Pharmacy Department, Jinan Huaiyin People’s Hospital, Jinan, China
| | - Junlin Wang
- Department of Pharmacy, Shandong University Second People’s Hospital, Jinan, China
| | - Guangqia Liu
- Department of Pharmacy, Jinan Licheng District Liubu Town Health Centre, Jinan, China
| | - Jiayin Meng
- Department of Pharmacy, Jinan Second People’s Hospital, Jinan, China
| | - Yingjie Zhang
- College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
48
|
Brandstetter J, Hoffmann L, Koopmann I, Schreiber T, Schulz B, Rosshart SP, Zechner D, Vollmar B, Kumstel S. Burrowing Behavior as Robust Parameter for Early Humane Endpoint Determination in Murine Models for Pancreatic Cancer. Animals (Basel) 2025; 15:1241. [PMID: 40362056 PMCID: PMC12071103 DOI: 10.3390/ani15091241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Due to late-stage diagnoses and limited treatment options, pancreatic cancer is predicted to be the second leading cause of cancer deaths by 2030. Many different murine models were developed over the past decades to test new therapies for this tumor entity. The laws and regulations demand the continuous refinement of animal experiments in order to improve animal welfare sustainably. A key aspect here is the definition of early humane endpoint criteria to avoid severe and lasting suffering of the animals through timely euthanasia. The present study analyzed the welfare of mice in different pancreatic cancer models, various mouse strains, and under different therapeutic interventions in preclinical testing. Their welfare was monitored before any intervention, during tumor progression, and on the days before the individual humane endpoint for each mouse by assessing body weight change, distress score, perianal temperature, burrowing behavior, nesting activity, and mouse grimace scale. The data was retrospectively analyzed via receiver operating characteristic curve analysis to quantify the predictability of each parameter for humane endpoint determination. Burrowing behavior proved to be a robust predictor of the humane endpoint two days in advance under various conditions, including diverse pancreatic cancer models and different therapeutic approaches.
Collapse
Affiliation(s)
- Jakob Brandstetter
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Lisa Hoffmann
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Ingo Koopmann
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Benjamin Schulz
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Stephan Patrick Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Simone Kumstel
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| |
Collapse
|
49
|
Zhang R, Danshiitsoodol N, Noda M, Yonezawa S, Kanno K, Sugiyama M. Stevia Leaf Extract Fermented with Plant-Derived Lactobacillus plantarum SN13T Displays Anticancer Activity to Pancreatic Cancer PANC-1 Cell Line. Int J Mol Sci 2025; 26:4186. [PMID: 40362423 PMCID: PMC12071683 DOI: 10.3390/ijms26094186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Pancreatic cancer is a highly malignant tumor that remains a significant global health burden. In this study, we demonstrated the anticancer potential of stevia leaf extract fermented with plant-derived Lactobacillus (L.) plantarum SN13T strain. Evaluation of antioxidant capacity (including DPPH and ABTS radical scavenging activities and H2O2-induced oxidative damage repair in HEK-293 cells), as well as cytotoxicity against pancreatic cancer cells (PANC-1) and non-cancerous human embryonic kidney (HEK-293), revealed that the fermented extract exhibited significantly enhanced antioxidant activity and cytotoxicity against PANC-1 cells while showing minimal toxicity to HEK-293 cells compared to the unfermented extract. Further, validation through clonogenic, migration, and wound-healing assays demonstrated that the fermented extract effectively inhibited the proliferation and migration of PANC-1 cells. The active compound in the fermented extract has been identified as chlorogenic acid methyl ester (CAME), with a concentration of 374.4 μg/mL. Flow cytometry analysis indicated that CAME significantly arrested PANC-1 cells in the G0/G1 phase and induced apoptosis. Furthermore, CAME upregulated the expression of pro-apoptotic genes Bax, Bad, Caspase-3/9, Cytochrome c, and E-cadherin, while downregulating the anti-apoptotic gene Bcl-2. These findings suggest that CAME exerts potent cytotoxic effects on PANC-1 cells by inhibiting cell proliferation and migration, arresting the cell cycle, and regulating apoptosis-related gene expression. In conclusion, stevia leaf extract fermented with L. plantarum SN13T, which contains CAME, may serve as a promising candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Rentao Zhang
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Sayaka Yonezawa
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| |
Collapse
|
50
|
Liu S, Su J, Zhao H, Bai R, Zeng L, Xue C, Deng S, Liu S, Chen Z, Xu Z, Zhou Y, Zhao S, Wu X, Peng X, Zhang J, Huang X, Zheng J, Zhao C, Zheng L. Identification of novel plasma proteins as promising noninvasive biomarker for early diagnosis and surveillance of pancreatic ductal adenocarcinoma. J Gastroenterol 2025:10.1007/s00535-025-02252-w. [PMID: 40285860 DOI: 10.1007/s00535-025-02252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Although cancer antigen 19-9 (CA 19-9) is the only FDA-approved biomarker for pancreatic ductal adenocarcinoma (PDAC), its diagnostic effectiveness is limited, as it may not be elevated in 15-25% of patients. This study aims to explore novel plasma proteins associated with PDAC as potential biomarkers for early diagnosis and clinical surveillance. METHODS Novel plasma protein biomarkers potentially causally associated with PDAC were identified using Mendelian randomization (MR). These biomarkers were validated in a multicenter study encompassing 230 tissue and 1,011 plasma samples to establish a diagnostic model for PDAC. Furthermore, their pre- and post-operative levels were compared to evaluate their potential as clinical surveillance biomarkers. RESULTS Genetically predicted expression of seven proteins potentially causally associated with an increased risk of PDAC. In a multicenter, large-scale study, Keratin 5 (KRT5) and Versican (VCAN) were identified as promising biomarkers for PDAC, with an area under the curve (AUC) of 0.90, and a combined panel including CA 19-9 achieved an AUC of 0.95. Additionally, plasma KRT5 and VCAN demonstrated superior diagnostic performance for early-stage PDAC with CA 19-9 levels below 37 U/mL (Stage I, AUC 0.85; Stage II, AUC 0.85). The specificity of plasma KRT5 and VCAN for PDAC was further validated by comparing their expression levels across various digestive cancers. Moreover, a significant decrease in plasma KRT5 and VCAN levels was observed post-surgery (P < 0.05), supporting their potential as biomarkers for clinical surveillance of PDAC. CONCLUSIONS Plasma KRT5 and VCAN proteins may serve as promising valuable biomarkers for the early diagnosis and clinical surveillance of PDAC.
Collapse
Affiliation(s)
- Shuang Liu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiachun Su
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hongzhe Zhao
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ruihong Bai
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lingxing Zeng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chunling Xue
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shuang Deng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shaoqiu Liu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ziming Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zilan Xu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yifan Zhou
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Sihan Zhao
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaoyu Wu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xinyi Peng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jialiang Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xudong Huang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jian Zheng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Chongyu Zhao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|